WO2016103155A1 - Triazolopyrimidine compounds and uses thereof - Google Patents

Triazolopyrimidine compounds and uses thereof Download PDF

Info

Publication number
WO2016103155A1
WO2016103155A1 PCT/IB2015/059843 IB2015059843W WO2016103155A1 WO 2016103155 A1 WO2016103155 A1 WO 2016103155A1 IB 2015059843 W IB2015059843 W IB 2015059843W WO 2016103155 A1 WO2016103155 A1 WO 2016103155A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
methyl
independently selected
pyrimidin
triazolo
Prior art date
Application number
PCT/IB2015/059843
Other languages
English (en)
French (fr)
Inventor
Ho Man Chan
Xiang-Ju Justin Gu
Ying Huang
Ling Li
Yuan Mi
Wei Qi
Martin Sendzik
Yongfeng SUN
Long Wang
Zhengtian Yu
Hailong Zhang
Ji Yue Zhang (Jeff)
Man ZHANG
Qiong Zhang
Kehao Zhao
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2015370524A priority Critical patent/AU2015370524B2/en
Priority to LTEP15820637.5T priority patent/LT3237418T/lt
Priority to JP2017533767A priority patent/JP6736559B2/ja
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to RS20190502A priority patent/RS58679B1/sr
Priority to NZ731664A priority patent/NZ731664A/en
Priority to CA2969090A priority patent/CA2969090C/en
Priority to EP15820637.5A priority patent/EP3237418B1/en
Priority to EA201791420A priority patent/EA032416B1/ru
Priority to TN2017000204A priority patent/TN2017000204A1/en
Priority to DK15820637.5T priority patent/DK3237418T3/da
Priority to MX2017008529A priority patent/MX2017008529A/es
Priority to KR1020177016880A priority patent/KR102534028B1/ko
Priority to UAA201704770A priority patent/UA120945C2/uk
Priority to BR112017010354A priority patent/BR112017010354B8/pt
Priority to CR20170285A priority patent/CR20170285A/es
Priority to SI201530725T priority patent/SI3237418T1/sl
Priority to MEP-2019-127A priority patent/ME03385B/me
Priority to ES15820637T priority patent/ES2722048T3/es
Priority to SG11201703880VA priority patent/SG11201703880VA/en
Priority to US15/538,348 priority patent/US10220036B2/en
Priority to CN201580070691.5A priority patent/CN107108637B/zh
Priority to MYPI2017701623A priority patent/MY186837A/en
Priority to PL15820637T priority patent/PL3237418T3/pl
Publication of WO2016103155A1 publication Critical patent/WO2016103155A1/en
Priority to IL252135A priority patent/IL252135B/en
Priority to PH12017501016A priority patent/PH12017501016B1/en
Priority to CONC2017/0005992A priority patent/CO2017005992A2/es
Priority to US16/246,380 priority patent/US20190142837A1/en
Priority to HRP20190805TT priority patent/HRP20190805T1/hr
Priority to CY20191100456T priority patent/CY1121901T1/el
Priority to US16/863,631 priority patent/US11207325B2/en
Priority to US17/505,271 priority patent/US11931363B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present disclosure relates to triazolopyrimidine compounds, compositions comprising such compounds, and their use for the treatment of Polycomb Repressive Complex 2 (PRC2)-mediated diseases or disorders.
  • PRC2 Polycomb Repressive Complex 2
  • Polycomb group (PcG) proteins are chromatin modifying enzymes that are dysregulated in many human cancers.
  • the Polycomb Repressive Complex 2 (PRC2) which includes SUZ12 (suppressor of zeste 12), EED (embryonic ectoderm
  • EZH2 enhanced of zeste homolog 2
  • PRC2 is the critical component of cellular machinery involved in the epigenetic regulation of gene transcription and plays critical function in development and tissue differentiation and regeneration.
  • EZH2 is the catalytic subunit, PRC2 requires at least EED and SUZ12 for its methyltransferase activity. EED, SUZ12 and EZH2 are overexpressed in many cancers, including but not limited to breast cancer, prostate cancer, hepatocellular carcinoma and etc.
  • EZH2 activating mutations have been identified in DLBCL (diffused large B cell lymphoma) patients and FL (follicular lymphoma) patients.
  • DLBCL diffuseused large B cell lymphoma
  • FL follicular lymphoma
  • PRC2 methyltransferase activity by compounds competing with the cofactor S-adenosyl methionine (SAM) in DLBCL reverses H3K27 methylation, re-activates expression of target genes and inhibits tumor growth/proliferation. Therefore, PRC2 provides a pharmacological target for DLBCL and other cancers.
  • SAM cofactor S-adenosyl methionine
  • R 1 , R 2 , R 3 , R 4 , R 5 , and n are as defined herein, including stereoisomers, tautomers, pharmaceutically acceptable salts, polymorphs, or solvates thereof, which are useful for the treatment of PRC2-mediated diseases or disorders.
  • the present disclosure also provides processes and intermediates for making the compounds of the present disclosure.
  • the present disclosure also provides pharmaceutical compositions comprising at least one of the compounds of the present disclosure and at least one pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition may further comprise at least one additional therapeutic agent.
  • additional therapeutic agents selected from: other anti-cancer agents, immunomodulators, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • the compounds of the present disclosure may be used in the treatment of diseases or disorders mediated by EED and/or PRC2.
  • the compounds of the present disclosure may be used in therapy.
  • the compounds of the present disclosure may be used for the manufacture of a medicament for the treatment of diseases or disorders mediated by EED and/or PRC2.
  • the present disclosure provides a method for the treatment of diseases or disorders mediated by EED and/or PRC2, comprising administering to a patient in need thereof a therapeutically effective amount of a first therapeutic agent optionally with a second therapeutic agent, wherein the first therapeutic agent is a compound of the present disclosure and the second therapeutic agent is one other type of therapeutic agent.
  • Examples of diseases or disorders mediated by EED and/or PRC2 include, but are not limited to, diffused large B cell lymphoma (DLBCL), follicular lymphoma, other lymphomas, leukemia, multiple myeloma, mesothelioma, gastric cancer, malignant rhabdoid tumor, hepatocellular carcinoma, prostate cancer, breast carcinoma, bile duct and gallbladder cancers, bladder carcinoma, brain tumors including neuroblastoma, schwannoma, glioma, glioblastoma and astrocytoma, cervical cancer, colon cancer, melanoma, endometrial cancer, esophageal cancer, head and neck cancer, lung cancer, nasopharyngeal carcinoma, ovarian cancer, pancreatic cancer, renal cell carcinoma, rectal cancer, thyroid cancers, parathyroid tumors, uterine tumors, and soft tissue sarcomas such as rhabdomyosarcom
  • the present disclosure provides a method for the treatment of diseases or disorders mediated by EED and/or PRC2, comprising administering to a patient in need thereof a therapeutically effective amount of a first therapeutic agent optionally with a second therapeutic agent, wherein the first therapeutic agent is an EED inhibitor and the second therapeutic agent is one other type of therapeutic agent; wherein the diseases or disorders are selected from diffused large B cell lymphoma (DLBCL), follicular lymphoma, other lymphomas, leukemia, multiple myeloma, gastric cancer, malignant rhabdoid tumor, and hepatocellular carcinoma.
  • DLBCL diffused large B cell lymphoma
  • follicular lymphoma other lymphomas
  • leukemia multiple myeloma
  • gastric cancer malignant rhabdoid tumor
  • hepatocellular carcinoma hepatocellular carcinoma
  • the compounds of the present disclosure can be used alone, in combination with other compounds of the present disclosure, or in combination with one or more, preferably one to two other agent(s), simultaneously or sequentially.
  • the present disclosure provides, inter alia, a compound of
  • R 1 and R 2 are independently H or halogen
  • R 3 is independently selected from: halogen, phenyl, and a 5- to 6-membered heteroaryl comprising carbon atoms and 1 -4 heteroatoms selected from N, NR a , O, and S(0) p ; wherein said phenyl and heteroaryl are substituted with 0-3 R 3A ;
  • each R 3C is independently selected from: C 3 -C 6 cycloalkyl, phenyl, and a 4- to 7- membered heterocycle comprising carbon atoms and 1 -4 heteroatoms selected from N, NR a , O, and S(0) p ; wherein each moiety is substituted with 0-2 R c ;
  • each R 3D is independently selected from: C C 4 alkyl and R 3C ;
  • R 3E and R 3G are, at each occurrence, independently selected from: H and C C 4 alkyl;
  • each R 3F is independently selected from: H and C C 4 alkyl substituted with 0-1
  • R 4 is independently selected from: H, halogen and C C 4 alkyl
  • R 5 is independently selected from OH and C C 4 alkyl
  • R b is independently selected from: halogen, OH and C C 4 alkoxy;
  • R d is independently selected from: OH and NR e R f ;
  • R e and R f are, at each occurrence, independently selected from: H and
  • each p is independently selected from 0, 1 and 2;
  • n and n are, at each occurrence, independently selected from 0 and 1 .
  • R 1 and R 2 are independently H or halogen
  • R 3 is independently selected from: halogen, phenyl, and a 5- to 6-membered heteroaryl comprising carbon atoms and 1 -4 heteroatoms selected from N, NR a , O, and S(0) p ; wherein said phenyl and heteroaryl are substituted with 0-3 R 3A ;
  • each R 3C is independently selected from: C 3 -C 6 cycloalkyl, phenyl, and a 4- to 7- membered heterocycle comprising carbon atoms and 1 -4 heteroatoms selected from N, NR a , O, and S(0) p ; wherein each moiety is substituted with 0-2 R c ;
  • each R 3D is independently selected from: C C 4 alkyl and R 3C ;
  • R 3E and R 3G are, at each occurrence, independently selected from: H and
  • each R 3F is independently selected from: H and C C 4 alkyl substituted with 0-1 R d ;
  • R 4 is independently selected from: H, halogen and C C 4 alkyl;
  • R 5 is independently C C 4 alkyl;
  • R b is independently selected from: halogen, OH and C 1 -C 4 alkoxy;
  • R d is independently selected from: OH and NR e R f ;
  • R e and R f are, at each occurrence, independently selected from: H and d-C 4 alkyl;
  • each p is independently selected from 0, 1 and 2;
  • n and n are, at each occurrence, independently selected from 0 and 1 .
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of the first aspect; wherein:
  • R 4 is H
  • n is independently selected from 0 and 1 ;
  • n 0.
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of the first or second aspect; wherein:
  • R 1 is independently H or F
  • R 2 is H
  • R 3 is independently selected from: phenyl and a 6-membered heteroaryl comprising carbon atoms and 1 -2 heteroatoms selected from N and NR a ; wherein said phenyl and heteroaryl are substituted with 0-3 R 3A .
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of any one of the first, second and third aspects; wherein:
  • R 3 is independently selected from: phenyl, pyridyl, pyrimidyl, pyridazinyl and pyrazinyl; wherein each moiety is substituted with 0-3 R 3A .
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of any of the first to fourth aspects, wherein:
  • R 3 is independently selected from:
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of any of the first to fifth aspects, wherein:
  • R 3B is independently selected from: OH, NH 2 , NH(C C 4 alkyl), N(d-d alkyl) 2 ,
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of any of the first to sixth aspects, wherein:
  • R 3 is independently selected from: ;
  • R is independently selected from: OH, N(C 1 -C 4 alkyl) 2 , d-d alkoxy,
  • R 1 is independently H or F
  • R 3A is independently selected from: F, CH 3 , -CH 2 OH, CH 2 F, CHF 2 , CF 3 , and
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of the first or second aspect; wherein:
  • R 1 is independently H or F
  • R 2 is H
  • R 3 is independently a 5-membered heteroaryl comprising carbon atoms and 1 -4 heteroatoms selected from N, NR a , O, and S(0) p ; wherein said heteroaryl is substituted with 0-3 R 3A ; and
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of any one of the first, second and tenth aspects; wherein:
  • R 3 is independently selected from: [0026]
  • the present disclosure includes a compound of Formula (I) or (IA), or a pharmaceutically acceptable salt thereof, within the scope of any one of the first, second and tenth aspects; wherein:
  • R is independently selected from: , , and
  • the present disclosure includes a compound of Formula (I) (IA), or (IA-1), or a pharmaceutically acceptable salt thereof, within the scope of any of the above aspects, wherein:
  • R 1 is F.
  • the present disclosure provides a compound selected from the exemplified examples or a pharmaceutically acceptable salt thereof, including all compounds of Examples 1 to 245.
  • the present disclosure provides a compound selected
  • Example 1 In another embodiment, provided is a compound of Example 1 or a pharmaceutically acceptable salt thereof, wherein the compound is 8-(1 ,3-dimethyl-1 H- pyrazol-5-yl)-N-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-[1 ,2,4]triazolo[4,3- c]pyrimidin-5-amine.
  • Example 1 In another embodiment, provided is a compound of Example 1 , wherein the compound is 8-(1 ,3-dimethyl-1 H-pyrazol-5-yl)-N-((5-fluoro-2,3-dihydrobenzofuran-4- yl)methyl)-[1 ,2,4]triazolo[4,3-c]pyrimidin-5-amine.
  • Example 2 In another embodiment, provided is a compound of Example 2 or a pharmaceutically acceptable salt thereof, wherein the compound is /V-((5-fluoro-2,3- dihydrobenzofuran-4-yl)methyl)-8-(2-methylpyridin-3-yl)-[1 ,2,4]triazolo[4,3-c]pyrimidin-5- amine.
  • Example 2 In another embodiment, provided is a compound of Example 2, wherein the compound is /V-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-8-(2-methylpyridin-3-yl)- [1 ,2,4]triazolo[4,3-c]pyrimidin-5-amine.
  • Example 5 In another embodiment, provided is a compound of Example 5 or a pharmaceutically acceptable salt thereof, wherein the compound is /V-((5-fluoro-2,3- dihydrobenzofuran-4-yl)methyl)-8-(6-methoxy-4-methylpyridin-3-yl)-[1 ,2,4]triazolo[4,3- c]pyrimidin-5-amine.
  • Example 5 In another embodiment, provided is a compound of Example 5, wherein the compound is /V-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-8-(6-methoxy-4- methylpyrid in-3-yl)-[1 ,2,4]triazolo[4,3-c]pyrimidin-5-amine.
  • Example 8 In another embodiment, provided is a compound of Example 8 or a pharmaceutically acceptable salt thereof, wherein the compound is 8-(2-cyclopropyl-4- methylpyrimidin-5-yl)-/V-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-[1 ,2,4]triazolo[4,3- c]pyrimidin-5-amine.
  • a compound of Example 8 wherein the compound is 8-(2-cyclopropyl-4-methylpyrimidin-5-yl)-/V-((5-fluoro-2,3- dihydrobenzofuran-4-yl)methyl)-[1 ,2,4]triazolo[4,3-c]pyrimidin-5-amine.
  • a compound of Example 207 or a pharmaceutically acceptable salt thereof wherein the compound is N-((5- fluorobenzofuran-4-yl)methyl)-8-(2-methylpyridin-3-yl)-[1 ,2,4]triazolo[4,3-c]pyrimidin-5- amine.
  • Example 207 provides a compound of Example 207, wherein the compound is N-((5-fluorobenzofuran-4-yl)methyl)-8-(2-methylpyridin-3-yl)- [1 ,2,4]triazolo[4,3-c]pyrimidin-5-amine.
  • Example 233 In another embodiment, provided is a compound of Example 233 or a pharmaceutically acceptable salt thereof, wherein the compound is /V-((5-fluoro-2,3- dihydrobenzofuran-4-yl)methyl)-8-(4-methyl-1 H-imidazol-1 -yl)-[1 ,2,4]triazolo[4,3- c]pyrimidin-5-amine.
  • Example 233 In another embodiment, provided is a compound of Example 233, wherein the compound is /V-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-8-(4-methyl-1 H-imidazol-1 - yl)-[1 ,2,4]triazolo[4,3-c]pyrimidin-5-amine.
  • the present disclosure provides a compound or a pharmaceutically acceptable salt thereof, selected from any subset list of compounds within the scope of the thirteenth aspect.
  • a compound or a pharmaceutically acceptable salt thereof selected from any subset list of compounds within the scope of the thirteenth aspect.
  • provided is the compound or a pharmaceutically acceptable salt thereof, within the scope of any one of the first to seventh aspects, wherein:
  • R 3 is independently ;
  • each R is independently selected from: C 1 -C 4 alkyl, C 1 -C 4 alkoxy,
  • R 3 is independently ;
  • R 3 is independently
  • each R 3A is independently selected from: C 1 -C 4 alkoxy, C C 4 haloalkoxy, and -0-C 3 -C 6 cycloalkyl.
  • R 3 is independently
  • each R 3A is independently selected from: OCH 3 , OCH 2 CH 3 , -OCHF 2 , and -O-cyclopropyl.
  • each R 3A is independently selected from: C 1 -C 4 alkoxy, C C 4 haloalkoxy, and -0-C 3 -C 6 cycloalkyl.
  • R 3 is independently ;
  • each R 3A is independently selected from: halogen, CN, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C C 4 haloalkyl, C C 4 haloalkoxy, N(C 1 -C 4 alkyl) 2 , and C 3 -C 6 cycloalkyl.
  • halogen CN, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, C C 4 haloalkyl, C C 4 haloalkoxy, N(C 1 -C 4 alkyl) 2 , and C 3 -C 6 cycloalkyl.
  • R 3 is independently ;
  • each R 3A is independently selected from: halogen, CN,
  • R 3B is independently selected from: OH, NH 2 , N(C 1 -C 4 alkyl) 2 , and
  • R 3c is independently selected from: C C 4 alkyl and 1 H-piperidin-4-yl.
  • the present disclosure includes a compound of Formula (IA), or a pharmaceutically acceptable salt thereof, within the scope of any one of the first, second, tenth, and eleventh aspects; wherein:
  • R J is independently selected from:
  • the present disclosure includes a compound of Formula (IA), or a pharmaceutically acceptable salt thereof, within the scope of any one of the first, second, tenth, and eleventh aspects; wherein:
  • R 3 is independently
  • each R 3A is independently selected from: C C 4 alkyl, C 1 -C 4 alkoxy,
  • R a is independently selected from: H, C C 4 alkyl substituted with 0-1 R b , and C 3 -C 6 cycloalkyl;
  • R b is independently selected from: OH and CrC 4 alkoxy.
  • the compounds of the present disclosure have IC50 values ⁇ 5 ⁇ , using the EED Alphascreen binding, LC-MS and/or ELISA assays disclosed herein, preferably, IC50 values ⁇ 1 ⁇ , more preferably, IC50 values ⁇ 0.5 ⁇ , even more preferably, IC50 values ⁇ 0.1 ⁇ . II. OTHER EMBODIMENTS
  • the present disclosure provides a composition comprising at least one of the compounds of the present disclosure or a
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising at least one of the compounds of the present disclosure or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable carrier, diluent or excipient.
  • the present disclosure provides a pharmaceutical composition, comprising a therapeutically effective amount of at least one of the compounds of the present disclosure or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition is useful in the treatment of diseases or disorders mediated by EED and/or PRC2.
  • the present disclosure provides a pharmaceutical composition as defined above further comprising additional therapeutic agent(s).
  • the present disclosure provides a process for making a compound of the present disclosure.
  • the present disclosure provides an intermediate for making a compound of the present disclosure.
  • the present disclosure provides a compound of the present disclosure, for use in therapy, alone, or optionally in combination with another compound of the present disclosure and/or at least one other type of therapeutic agent.
  • the present disclosure provides a compound of the present disclosure for use in therapy, for the treatment of diseases or disorders mediated by EED and/or PRC2, alone, or optionally in combination with another compound of the present disclosure and/or at least one other type of therapeutic agent.
  • the present disclosure provides a method for the treatment of diseases or disorders mediated by EED and/or PRC2, comprising administering to a patient in need of such treatment a therapeutically effective amount of at least one of the compounds of the present disclosure, alone, or optionally in combination with another compound of the present disclosure and/or at least one other type of therapeutic agent.
  • the present disclosure provides a method for the treatment of diseases or disorders mediated by EED and/or PRC2, comprising administering to a patient in need thereof a therapeutically effective amount of a first and second therapeutic agent, wherein the first therapeutic agent is a compound of the present disclosure and the second therapeutic agent is one other type of therapeutic agent.
  • the present disclosure also provides the use of a compound of the present disclosure for the manufacture of a medicament for the treatment of diseases or disorders mediated by EED and/or PRC2, alone, or optionally in combination with another compound of the present disclosure and/or at least one other type of therapeutic agent.
  • the present disclosure provides a combined preparation of a compound of the present disclosure and additional therapeutic agent(s) for use in therapy.
  • the present disclosure provides a combination of a compound of the present disclosure and additional therapeutic agent(s) for simultaneous or separate use in therapy.
  • the present disclosure provides a combined preparation of a compound of the present disclosure and additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment of diseases or disorders mediated by EED and/or PRC2.
  • the compound may be administered as a
  • Examples of diseases or disorders mediated by EED and/or PRC2 include, but are not limited to, diffused large B cell lymphoma (DLBCL), follicular lymphoma, other lymphomas, leukemia, multiple myeloma, mesothelioma, gastric cancer, malignant rhabdoid tumor, hepatocellular carcinoma, prostate cancer, breast carcinoma, bile duct and gallbladder cancers, bladder carcinoma, brain tumors including neurobalstoma, glioma, glioblastoma and astrocytoma, cervical cancer, colon cancer, melanoma, endometrial cancer, esophageal cancer, head and neck cancer, lung cancer, nasopharhyngeal carcinoma, ovarian cancer, pancreatic cancer, renal cell carcinoma, rectal cancer, thyroid cancers, parathyroid tumors, uterine tumors, and soft tissue sarcomas selected from rhabdomyosarcoma (RM
  • the present disclosure provides a method for the treatment of diseases or disorders mediated by EED and/or PRC2, comprising administering to a patient in need thereof a therapeutically effective amount of a first optionally with a second therapeutic agent, wherein the first therapeutic agent is an EED inhibitor and the second therapeutic agent is one other type of therapeutic agent;
  • diseases or disorders are selected from diffused large B cell lymphoma (DLBCL), follicular lymphoma, other lymphomas, leukemia, multiple myeloma, gastric cancer, malignant rhabdoid tumor, and hepatocellular carcinoma.
  • DLBCL diffused large B cell lymphoma
  • follicular lymphoma other lymphomas
  • leukemia multiple myeloma
  • gastric cancer malignant rhabdoid tumor
  • hepatocellular carcinoma hepatocellular carcinoma
  • additional therapeutic agent(s) used in combined pharmaceutical compositions or combined methods or combined uses are selected from one or more, preferably one to three, of the following therapeutic agents: other anticancer agents, immunomodulators, anti-allergic agents, anti-nausea agents (or antiemetics), pain relievers, cyto protective agents, and combinations thereof.
  • heteroatoms refers to nitrogen (N), oxygen (O) or sulfur (S) atoms, in particular nitrogen or oxygen.
  • N nitrogen
  • O oxygen
  • S sulfur
  • any heteroatom with unsatisfied valences is assumed to have hydrogen atoms sufficient to satisfy the valences.
  • alkyl refers to a hydrocarbon radical of the general formula C n H 2n +i .
  • the alkane radical may be straight or branched.
  • Ci-C 10 alkyl or C ⁇ to C 10 alkyl refers to a monovalent, straight, or branched aliphatic group containing 1 to 10 carbon atoms (e.g., methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, s-butyl, t-butyl, n-pentyl, 1 -methylbutyl, 2-methylbutyl, 3-methylbutyl, neopentyl, 3,3-dimethylpropyl, hexyl, 2-methylpentyl, heptyl, and the like).
  • alkylene refers to a divalent alkyl group.
  • C -C 6 alkylene or “C ⁇ to C 6 alkylene” refers to a divalent, straight, or branched aliphatic group containing 1 to 6 carbon atoms (e.g., methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), n-propylene (-CH 2 CH 2 CH 2 -), iso-propylene (-CH(CH 3 )CH 2 -), n-butylene, sec-butylene, iso-butylene, tert-butylene, n-pentylene, isopentylene, neopentylene, n-hexylene and the like).
  • alkoxy refers to an alkyl linked to an oxygen, which may also be represented as -O-R or -OR, wherein the R represents the alkyl group.
  • C -C 6 alkoxy or “C ⁇ to C 6 alkoxy” is intended to include C 2 , C 3 , C 4 , C 5 , and C 6 alkoxy groups.
  • Example alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (e.g. , n-propoxy and isopropoxy), and f-butoxy.
  • alkylthio or “thioalkoxy” represents an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example methyl-S- and ethyl-S-.
  • Halogen or "halo” may be fluorine, chlorine, bromine or iodine (preferred halogens as substituents are fluorine and chlorine).
  • Haloalkyi is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with one or more halogens.
  • haloalkyi include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl, heptafluoropropyl, and heptachloropropyl.
  • haloalkyi also include "fluoroalkyl” that is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with one or more fluorine atoms.
  • Haloalkoxy represents a haloalkyi group as defined above with the indicated number of carbon atoms attached through an oxygen bridge.
  • C -C 6 haloalkoxy or “ ⁇ to C 6 haloalkoxy” is intended to include Ci , C 2 , C 3 , C 4 , C 5 , and C 6 haloalkoxy groups.
  • Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluorothoxy.
  • haloalkylthio or “thiohaloalkoxy” represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example
  • oxo or -C(O)- refers to a carbonyl group.
  • a ketone, aldehyde, or part of an acid, ester, amide, lactone, or lactam group for example, a ketone, aldehyde, or part of an acid, ester, amide, lactone, or lactam group.
  • cycloalkyl refers to nonaromatic carbocyclic ring that is fully hydrogenated ring, including mono-, bi- or poly-cyclic ring systems.
  • C 3 -C 8 cycloalkyl or " C 3 to C 8 cycloalkyl " is intended to include C 3 , C 4 , C 5 , C 6 , C 7 and C 8 cycloalkyl groups.
  • Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbornyl.
  • aryl refers to 6- to 10-membered aromatic carbocyclic moieties having a single (e.g., phenyl) or a fused ring system (e.g., naphthalene.).
  • a typical aryl group is phenyl group.
  • benzyl refers to a methyl group on which one of the hydrogen atoms is replaced by a phenyl group.
  • R is hydrogen, C ⁇ alkyl or a nitrogen protecting group (for example, carbobenzyloxy, p-methoxybenzyl carbonyl, t-butoxycarbonyl, acetyl, benzoyl, benz
  • a 3 to 8 membered heterocycloalkyl includes epoxy, aziridinyl, azetidinyl, imidazolidinyl, pyrazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydrothienyl 1 ,1 -dioxide, oxazolidinyl, thiazolidinyl, pyrrolidinyl, pyrrolidinyl-2-one, morpholino, piperazinyl, piperidinyl, piperidinylone, pyrazolidinyl, hexahydropyrimidinyl, 1 ,4-dioxa-8-aza-spiro[4.5]dec-8-yl, thiomorpholino,
  • Partially saturated heterocyclic rings include groups such as
  • a partially saturated heterocyclic ring also includes groups wherein the heterocyclic ring is fused to an aryl or heteroaryl ring (e.g., 2,3-dihydrobenzofuranyl, indolinyl (or 2,3-dihydroindolyl), 2,3- dihydrobenzothiophenyl, 2,3-dihydrobenzothiazolyl, 1 ,2,3,4-tetrahydroquinolinyl, 1 ,2,3,4- tetrahydroisoquinolinyl, 5,6,7,8-tetrahydropyrido[3,4-b]pyrazinyl, and the like).
  • aryl or heteroaryl ring e.g., 2,3-dihydrobenzofuranyl, indolinyl (or 2,3-dihydroindolyl), 2,3- dihydrobenzothiophenyl, 2,3-dihydrobenzothiazolyl, 1 ,2,3,4
  • partially or fully saturated heterocycle refers to a nonaromatic ring that is either partially or fully hydrogenated and may exist as a single ring, bicyclic ring (including fused rings) or a spiral ring.
  • the heterocyclic ring is generally a 3- to 12-membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen.
  • heterocycloalkyl and “partially saturated heterocycle”.
  • spiral rings include 2,6-diazaspiro[3.3]heptanyl, 3-azaspiro[5.5]undecanyl, 3,9-diazaspiro[5.5]undecanyl, and the like.
  • heteroaryl refers to aromatic moieties containing at least one heteroatom (e.g., oxygen, sulfur, nitrogen or combinations thereof) within a 5- to 10- membered aromatic ring system (e.g., pyrrolyl, pyridyl, pyrazolyl, indolyl, indazolyl, thienyl, furanyl, benzofuranyl, oxazolyl, isoxazolyl, imidazolyl, triazolyl, tetrazolyl, triazinyl, pyrimidinyl, pyrazinyl, thiazolyl, purinyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, benzopyranyl, benzothiophenyl, benzoimidazolyl, benzoxazolyl, 1 H- benzo[d][1 ,2,3]triazolyl, and the like.).
  • the heteroaromatic moiety may consist of a single or fused ring system.
  • a typical single heteroaryl ring is a 5- to 6-membered ring containing one to four heteroatoms independently selected from oxygen, sulfur and nitrogen and a typical fused heteroaryl ring system is a 9- to 10-membered ring system containing one to four heteroatoms independently selected from oxygen, sulfur and nitrogen.
  • the fused heteroaryl ring system may consist of two heteroaryl rings fused together or a heteroaryl fused to an aryl (e.g., phenyl).
  • heterocycloalkyl partially or fully saturated heterocycle
  • partially saturated heterocycle partially saturated heterocycle
  • heteroaryl partially saturated heterocycle
  • a dotted ring When a dotted ring is used within a ring structure, this indicates that the ring structure may be saturated, partially saturated or unsaturated.
  • substituted means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that normal valencies are maintained and that the substitution results in a stable compound.
  • 2 hydrogens on the atom are replaced.
  • Keto substituents are not present on aromatic moieties.
  • nitrogen atoms e.g. , amines
  • these may be converted to N-oxides by treatment with an oxidizing agent (e.g. , mCPBA and/or hydrogen peroxides) to afford other compounds of this disclosure.
  • an oxidizing agent e.g. , mCPBA and/or hydrogen peroxides
  • shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxide (N ⁇ 0) derivative.
  • any variable occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence.
  • a group is shown to be substituted with 0-3 R groups, then said group may be unsubstituted or substituted with up to three R groups, and at each occurrence R is selected independently from the definition of R.
  • R 3A in the R 3 definition such that when R 3 is phenyl or 5- to 6-membered heteroaryl, these groups are either unsubstituted (not substituted with R 3A ) or substituted with one, two or three R 3A groups which are independently selected at each occurrence from the given definitions for R 3A .
  • R 3 is phenyl or 5- to 6-membered heteroaryl
  • R 3A is phenyl or 5- to 6-membered heteroaryl
  • this disclosure is intended to cover all possible tautomers even when a structure depicts only one of them.
  • phrases "pharmaceutically acceptable” indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • the term "compounds of the present invention” or “compounds of the present disclosure” refers to compounds of Formula (I), (IA) or (IA-1), as well as isomers, such as stereoisomers (including diastereoisomers, enantiomers and racemates), geometrical isomers, conformational isomers (including rotamers and astropisomers), tautomers, isotopically labeled compounds (including deuterium substitutions), and inherently formed moieties (e.g., polymorphs, solvates and/or hydrates). When a moiety is present that is capable of forming a salt, then salts are included as well, in particular pharmaceutically acceptable salts.
  • Enantiomers are a pair of stereoisomers that are non- superimposable mirror images of each other.
  • a 1 :1 mixture of a pair of enantiomers is a "racemic” mixture. The term is used to designate a racemic mixture where appropriate.
  • a single stereoisomer with known relative and absolute configuration of the two chiral centers is designated using the conventional RS system (e.g., (1 S.2S)); a single stereoisomer with known relative configuration but unknown absolute configuration is designated with stars (e.g., (1 R*,2R*)); and a racemate with two letters (e.g, (1 RS.2RS) as a racemic mixture of (1 R.2R) and (1 S.2S); (1 RS.2SR) as a racemic mixture of (1 R.2S) and (1 S.2R)).
  • the conventional RS system e.g., (1 S.2S
  • stars e.g., (1 R*,2R*
  • a racemate with two letters e.g, (1 RS.2RS) as a racemic mixture of (1 R.2R) and (1 S.2S
  • (1 RS.2SR a racemic mixture of (1 R.2S) and (1 S.2R
  • Diastereoisomers are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other.
  • the absolute stereochemistry is specified according to the Cahn- Ingold- Prelog R-S system. When a compound is a pure enantiomer the stereochemistry at each chiral carbon may be specified by either R or S.
  • Resolved compounds whose absolute configuration is unknown can be designated (+) or (-) depending on the direction (dextro- or levorotatory) which they rotate plane polarized light at the wavelength of the sodium D line. Alternatively, the resolved compounds can be defined by the respective retention times for the corresponding
  • Geometric isomers may occur when a compound contains a double bond or some other feature that gives the molecule a certain amount of structural rigidity. If the compound contains a double bond , the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans-configuration .
  • Conformational isomers are isomers that can differ by rotations about one or more a bonds. Rotamers are conformers that differ by rotation about only a single a bond.
  • atropisomer refers to a structural isomer based on axial or planar chirality resulting from restricted rotation in the molecule.
  • Optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques (e.g. , separated on chiral SFC or HPLC chromatography columns, such as CHIRALPAK® and CHIRALCEL® available from DAICEL Corp. using the appropriate solvent or mixture of solvents to achieve good separation).
  • the present compounds can be isolated in optically active or racemic forms.
  • Optically active forms may be prepared by resolution of racemic forms or by synthesis from optically active starting materials. All processes used to prepare compounds of the present disclosure and intermediates made therein are considered to be part of the present disclosure. When enantiomeric or diastereomeric products are prepared, they may be separated by conventional methods, for example, by chromatography or fractional crystallization.
  • the end products of the present disclosure are obtained either in free (neutral) or salt form. Both the free form and the salts of these end products are within the scope of the disclosure. If so desired, one form of a compound may be converted into another form. A free base or acid may be converted into a salt; a salt may be converted into the free compound or another salt; a mixture of isomeric compounds of the present disclosure may be separated into the individual isomers. [00112] Pharmaceutically acceptable salts are preferred. However, other salts may be useful, e.g. , in isolation or purification steps which may be employed during preparation, and thus, are contemplated within the scope of the disclosure.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • pharmaceutically acceptable salts include, but are not limited to, acetate, ascorbate, adipate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, caprate,
  • chloride/hydrochloride chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, glutamate, glutarate, glycolate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate/hydroxymalonate, mandelate, mesylate, methylsulphate, mucate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phenylacetate, phosphate/hydrogen phosphate/dihydrogen phosphate,
  • subsalicylate, tartrate, tosylate, trifluoroacetate or xinafoate salt form
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids.
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, preferably hydrochloric acid.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
  • the pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Allen, L.V., Jr., ed.,
  • Compounds of the invention that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co-crystals with suitable co-crystal formers.
  • These co-crystals may be prepared from compounds of the present invention by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of the present invention with the co-crystal former under crystallization conditions and isolating co- crystals thereby formed.
  • Suitable co-crystal formers include those described in WO 2004/078163. Hence the invention further provides co-crystals comprising a compound of the present invention.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 F 31 P, 32 P, 35 S, 36 CI, 125 l respectively.
  • the present disclosure includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3 H, 13 C, and 14 C , are present.
  • Such isotopically labelled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F or labeled compound may be particularly desirable for PET or SPECT studies.
  • Isotopically labeled compounds of this present disclosure can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • substitution with heavier isotopes may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index.
  • deuterium in this context is regarded as a substituent of a compound of the present disclosure.
  • concentration of such a heavier isotope, specifically deuterium may be defined by the isotopic enrichment factor.
  • isotopic enrichment factor as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium
  • such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium
  • Isotopically-labeled compounds of the present disclosure can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
  • Such compounds have a variety of potential uses, e.g. , as standards and reagents in determining the ability of a potential pharmaceutical compound to bind to target proteins or receptors, or for imaging compounds of this disclosure bound to biological receptors in vivo or in vitro.
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. It is preferred that compounds of the present disclosure do not contain a N-halo, S(0)2H, or S(0)H group.
  • solvate means a physical association of a compound of this disclosure with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • the solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement.
  • the solvate may comprise either a stoichiometric or nonstoichiometric amount of the solvent molecules.
  • “Solvate” encompasses both solution-phase and isolable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.
  • polymorph(s) refer to crystalline form(s) having the same chemical structure/composition but different spatial arrangements of the molecules and/or ions forming the crystals.
  • Compounds of the present disclosure can be provided as amorphous solids or crystalline solids. Lyophilization can be employed to provide the compounds of the present disclosure as a solid.
  • EED refers to the protein product of the gene embryonic ectoderm development.
  • PRC2 refers to Polycomb Repressive Complex 2.
  • PRC2-mediated disease or disorder refers to any disease or disorder which is directly or indirectly regulated by PRC2. This includes, but is not limited to, any disease or disorder which is directly or indirectly regulated by EED.
  • diseases or disorders mediated by EED and/or PRC2 refers to diseases or disorders which are directly or indirectly regulated by EED and/or PRC2.
  • patient encompasses all mammalian species.
  • the term "subject” refers to an animal. Typically the animal is a mammal. A “subject” also refers to any human or non-human organism that could potentially benefit from treatment with an EED inhibitor. A subject also refers to for example, primates (e.g. , humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human. Exemplary subjects include human beings of any age with risk factors for cancer disease.
  • a subject is "in need of a treatment if such subject would benefit biologically, medically or in quality of life from such treatment (preferably, a human).
  • the term “inhibit”, “inhibition” or “inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • the term "treat', "treating” or “treatment” of any disease/ disorder refers to the treatment of the disease/disorder in a mammal, particularly in a human, and includes: (a) ameliorating the disease/disorder, (i.e. , slowing or arresting or reducing the development of the disease/disorder, or at least one of the clinical symptoms thereof); (b) relieving or modulating the disease/disorder, (i.e., causing regression of the disease/disorder, either physically, (e.g.
  • stabilization of a discernible symptom physiologically, (e.g., stabilization of a physical parameter), or both); (c) alleviating or ameliorating at least one physical parameter including those which may not be discernible by the subject; and/or (d) preventing or delaying the onset or development or progression of the disease or disorder from occurring in a mammal, in particular, when such mammal is predisposed to the disease or disorder but has not yet been diagnosed as having it.
  • preventing or “prevention” cover the preventive treatment (i.e., prophylaxis and/or risk reduction) of a subclinical disease-state in a mammal, particularly in a human, aimed at reducing the probability of the occurrence of a clinical disease-state.
  • Patients are selected for preventative therapy based on factors that are known to increase risk of suffering a clinical disease state compared to the general population.
  • "Prophylaxis” therapies can be divided into (a) primary prevention and (b) secondary prevention.
  • Primary prevention is defined as treatment in a subject that has not yet presented with a clinical disease state, whereas secondary prevention is defined as preventing a second occurrence of the same or similar clinical disease state.
  • risk reduction covers therapies that lower the incidence of development of a clinical disease state.
  • primary and secondary prevention therapies are examples of risk reduction.
  • “Therapeutically effective amount” is intended to include an amount of a compound of the present disclosure that will elicit the biological or medical response of a subject, for example, reduction or inhibition of EED and/or PRC2, or ameliorate symptoms, alleviate conditions, slow or delay disease progression, or prevent a disease or disorder mediated by PRC2.
  • the term refers to combined amounts of the active ingredients that result in the preventive or therapeutic effect, whether administered in combination, serially, or simultaneously.
  • the compounds of the present disclosure can be prepared in a number of ways known to one skilled in the art of organic synthesis in view of the methods, reaction schemes and examples provided herein.
  • the compounds of the present disclosure can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art. Preferred methods include, but are not limited to, those described below.
  • the reactions are performed in a solvent or solvent mixture appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the disclosure
  • the starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wis.) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1 -19, Wiley, New York (1967-1999 ed.), Larock, R.C., Comprehensive Organic Transformations, 2 nd -ed., Wley-VCH Weinheim, Germany (1999), or Beilsteins Handbuch der organischen
  • reaction schemes depicted below provide potential routes for synthesizing the compounds of the present disclosure as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • Scheme 1 describes potential routes for producing the compounds of the present disclosure which include compounds of Formula (IA).
  • Compounds of Formula (IA) can be made substantially optically pure by either using substantially optically pure starting material or by separation chromatography, recrystallization or other separation techniques well-known in the art. For a more detailed description, see the Example section below.
  • Reverse phase preparative HPLC was carried out using C18 columns with UV 214 nm and 254 nm or prep LC-MS detection eluting with gradients of Solvent A (water with 0.1 % TFA) and Solvent B (acetonitrile with 0.1 % TFA) or with gradients of Solvent A (water with 0.05% TFA) and Solvent B (acetonitrile with 0.05% TFA) or with gradients of Solvent A (water with 0.05% ammonia) and Solvent B ( acetonitrile with 0.05% ammonia).
  • Reverse phase analytical HPLC/MS was performed on Agilent LC1200 systems coupled with 61 10 (Methods A-D), or 6120 (Method E and F), or 6130 (Method G) Mass Spectrometer.
  • Method A Linear gradient of 5% to 95% B over 1.2 min, with 1 min hold at 95% B; UV visualization at 214 nm and 254 nm
  • Solvent A 0.1 % trifluoroacetic acid, 99.9% water
  • Solvent B 0.1 % trifluoroacetic acid, 99.9% acetonitrile.
  • Method B Linear gradient of 5% to 95% B over 1.5 min, with 1 min hold at 95% B;
  • Solvent A water with 10mM Ammonium hydrogen carbonate
  • Solvent B acetonitrile
  • Solvent A 0.1 % trifluoroacetic acid, 99.9% water
  • Solvent B 0.1 % trifluoroacetic acid, 99.9% acetonitrile.
  • Method D Linear gradient of 5% to 95% B over 1.4 min, with 1.6 min hold at 95% B,
  • Solvent A water with 10mM Ammonium hydrogen carbonate
  • Solvent B acetonitrile
  • Method E Linear gradient of 5% to 95% B over 1.5 min, with 1 min hold at 95% B;
  • Solvent A water with 10mM Ammonium hydrogen carbonate
  • Solvent B acetonitrile
  • Method F Linear gradient of 5% to 95% B over 1 .5 min, with 1 min hold at 95% B;
  • Solvent A water with 0.1 % ammonia
  • Solvent B acetonitrile.
  • Method G Linear gradient of 10% to 95% B over 2 min, with 1 min hold at 95% B;
  • UV visualization at 214 nm, 254 nm and 300 nm
  • Solvent B MeOH with 0.1 % formic acid.
  • starting materials are generally available from a non-excluding commercial sources such as TCI Fine Chemicals (Japan), Shanghai Chemhere Co., Ltd. (Shanghai, China), Aurora Fine Chemicals LLC (San Diego, CA), FCH Group (Ukraine), Aldrich Chemicals Co. (Milwaukee, Wis.), Lancaster Synthesis, Inc. (Windham, N.H.), Acros Organics (Fairlawn, N.J.), Maybridge Chemical Company, Ltd. (Cornwall, England), Tyger Scientific (Princeton, N.J.), AstraZeneca
  • Methyl 2-methylbenzofuran-7-carboxylate (A3.2): A mixture of A3.1 (1 .0 g, 5.26 mmol) and cesium fluoride (1 .038 g, 6.84 mmol) in N,N-diethylaniline (5 mL, 5.26 mmol) was irritated by microwave at 200 °C for 30 min. After diluted with ehter insoluble materails were removed by decantation. The crude products were seperated on column chromatography by using a mixed solvent of hexane and ethyl acetate(10:1 , v/v) to give the title compound (500 mg, 50%).
  • tert-Butyl 4-(5-bromopyridin-2-yl)-3-oxopiperazine-1-carboxylate (B16.1): A mixture of 2,5-dibromopyridine (1 .0 g, 4.21 mmol), pyrrolidin-2-one (2.54 g, 12.7 mmol), K 2 C0 3 (1 .16 g, 8.42 mmol) , Cul (40 mg , 0.21 mmol), N1 ,N1 ,N2,N2-tetramethylethane- 1 ,2-diamine (92 mg , 0.63 mmol) and dioxane (10 mL) was stirred at 1 10 °C for 12 h.
  • B24.1 4-(5-bromo-3-methylpyridin-2-yl)morpholine (B24.1): A mixture of 5-bromo-2- fluoro-3-methylpyridine (2.5 g, 13.2 mmol), morpholine (3.4 g, 39.6 mmol), K 2 C0 3 (5.5 g, 39.6 mmol) in 40 mL DMSO was heated to 120 °C and stirred for 16 h. The mixture was cooled to rt. 200 mL water was added, extracted with ethyl acetate (150 mL ⁇ 3).
  • Methyl(m-tolyl)sulfane (B29.1): To a mixture of 3-methylbenzenethiol (2 g, 16 mmol) in 20 mL of DMF was added NaH (0.96 g, 24 mmol) at 0 °C. The mixture was stirred for 30 min at 25 °C. After coolig to 0 °C, CH 3 I (22.7 g, 160 mmol) was added dropwise. The mixture was stirred for 2 h at rt, diluted with 100 mL water, extracted with ethyl acetate (60 mL ⁇ 3).
  • (E)-Ethyl 3-(5-bromopyridin-2-yl)acrylate (B30.1): A mixture of 5- bromopicolinaldehyde (0.93 g, 5 mmol), ethyl 2-bromoacetate (1 .25 g, 7.5 mmol), NaHC0 3 (1 .26 g, 15 mmol), PPh 3 (1 .83 g, 7 mmol) , water (10 mL) in 5 mL ethyl acetate was stirred for 16 h at 25 °C under N 2 atmosphere.
  • LC-MS: [M+H] + 260.0.
  • Example 2 was prepared as follows. To a suspension of A1 (25.5 g, 70 mmol), 2-methyl-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridine (30.6 g, 140 mmol) and NaHC0 3 (35.3 g, 420 mmol) in a mixture solution of 1 ,4-dioxane (300 ml_) and H 2 0 (100 ml_) was added PdCI 2 (dppf) (5.94 g, 612 mmol). The mixture was degassed with N 2, heated at 1 10 °C for 1 h. The resulting mixture was cooled to rt and conentrated under reduced pressure.
  • A1 25.5 g, 70 mmol
  • 2-methyl-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridine 30.6 g, 140 mmol
  • NaHC0 3 35.3 g, 420 m
  • Example 2 (13.6 g, 52%) 1 H-NMR (500 MHz, DMSO-c/ 6 ) ⁇ ppm 2.40 (s, 3H), 3.33 (t, 2H), 4.56 (t, 2H), 4.72 (s, 2H), 6.72 (dd, 1 H), 6.96 (dd, 1 H), 7.31 (dd, 1 H), 7.66 (s, 1 H), 7.74 (d, 1 H), 8.51 (d, 1 H), 8.72 (t, 1 H), 9.49 (s, 1 H).
  • LC-MS: [M+H] + 376.9.
  • Example 2 To a suspension of Example 2 (6.0 g, 15.94 mmol) in 100 mL of IPA, a solution of 0.5 N HCI in IPA (33.0 mL, 16.50 mmol) was added dropwise at rt. The suspension was stirred at 50 °C for 12 h, then cooled to rt and stirred for 5 h.
  • Example 7 To a mixture 7.1 (30 mg, 0.08 mmol) in THF (3ml_) was added TBAF (0.6 mL, 0.6 mmol) and stirred for 4 h. The mixture was concentrated and purified by Prep-HPLC to afford the title compound (20 mg, 87%) as a white solid.
  • N-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-8-(pyridin-3-yl)- [1,2,4]triazolo[4,3-c]pyrimidin-5-amine (10.1): The title compound was prepared by using a procedure similar to that of Example 1 by replacing 1 ,3-dimethyl-1 H-pyrazol-5- ylboronic acid with 3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridine.
  • Example 10 To a mixture of 10.1 (1 10mg, 0.3mmol ) in CHCI 3 (5ml_) was added mCPBA (163mg, 0.6mmol).
  • reaction mixture was purged with N 2 for 3 times, then stirred at 90 °C for 1 h.
  • the mixture was filtered, the solid was washed with DMSO (2 mL).
  • the filtrate was concentrated and purified by prep-HPLC (NH 4 HC0 3 ) to give the title comppound (13 mg, 48%) as a white solid.
  • EED As a key component of PRC2 complex, EED has no intrinsic enzymatic activity. However, it is critical for proper PRC2 function. EED directly binds to H3K27me3 and this binding event localizes the PRC2 complex to the chromatin substrate and allosterically activates the methyltransferase activity. Targeting the allosteric site within the regulatory EED subunit of PRC2, may offer a novel and unique angle to be advantageous to, or complementary to, directly targeting the SAM competition mechanism of EZH2 or PRC2. Therefore, targeting EED represents a highly attractive strategy for the development of a novel therapy for the treatment of many forms of cancers. In particular, the need exists for small molecules that inhibit the activity of PRC2 through targeting EED. It has now been found that triazolopyrimidine derivatives as presently disclosed are useful to target EED for the treatment of EED or PRC2- mediated diseases or disorders, especially cancers.
  • the utility of the compounds of the present invention may be demonstrated using any one of the following test procedures.
  • Compounds of the present disclosure were assessed for their ability to inhibit PRC2 activity in a pentameric complex of EZH2, SUZ12, EED, Rbap48 and AEBP in biochemical assays.
  • the ability of compounds of the present disclosure to inhibit cellular activity of PRC2 was assessed by analysing histone H3 lysine 27 methylation in human cell lines.
  • the ability of compounds of the present disclosure to inhibit cancers was derived from their ability to modulate activity in human cancer cell lines bearing specific dependence to PRC2 activity to maintain cancerous growth.
  • AlphaScreen detection beads mix was prepared immediately before use by mixing nickel chelate acceptor beads and streptavidin donor beads in a 1 :1 ratio (Perkin Elmer, Product No.6760619C/M/R) into the buffer described above. Then 4 ⁇ _ of detection beads mix was added to the plate and incubate in the dark at the rt for 1 h. The final concentration of donor and acceptor beads was 10 ⁇ g/mL for each. Plates were read on EnVision (PerkinElmer) using the AlphaScreen setting adapted for optimal signal detection with a 615 nm filter, after sample excitation at 680 nm. The emission signal at 615 nm was used to quantify compounds inhibition.
  • AlphaScreen signals were normalized based on the reading coming from the positive (maximum signal control) and negative controls (minimum signal control) to give percentage of activities left. The data were then fit to a dose response equation using the program Helios (Novartis) to get the IC50 values.
  • Helios is a Novartis in-house assay data analysis software using the methods described by Normolle, D. P., Statistics in Medicine, 12:2025-2042 (1993); Formenko, I. et al, Computer Methods and Programs in Biomedicine, 82, 31 -37 (2006); Sebaugh, J. L, Pharmaceutical Statistics, 10:128-134 (201 1); Kelly, C.
  • AlphaScreen beads Compounds were diluted as described in the preceding section, and the assay was performed by adding 12 ⁇ of 10 nM biotin-miniPEG-His6 peptide in the above buffer and incubating for 20 min at rt prior to addition of the beads to10 ⁇ g/mL each. The plates were then incubated for 1 h at rt in dark before being read on EnVison. EED LC-MS Assay
  • Representative compounds of the present disclosure were serially and separately diluted 3-fold in DMSO to obtain a total of eight or twelve concentrations. Then the test compounds at each concentration (120 nl_ of each) were transferred by Mosquito into a 384-well Perkin Elmer ProxiPlate 384 plus plates. Solutions (6 ⁇ _) of 24 nM the wild type PRC2 (wtPRC2) complex and 2 ⁇ SAM in reaction buffer (20 mM Tris, pH 8.0, 0.1 % BSA, 0.01 % Triton, 0.5 mM DTT) were added to the wells that were then incubated with the test compound for 20 min. A 6 ⁇ _ solution of 3 ⁇ of the peptide substrate
  • H3K27MeO histone H3[21 -44]-biotin
  • reaction buffer was added to initiate each reaction.
  • the final components in the reaction solution include 12 nM wtPRC2 complex, 1 ⁇ SAM, and 1 .5 ⁇ H3K27meO peptide with varying concentration of the compounds.
  • a positive control consisted of the enzyme, 1 ⁇ SAM and 1 .5 ⁇ substrate in the absence of the test compound, and a negative control consisted of 1 ⁇ SAM and 1 .5 ⁇ substrate only.
  • Each reaction was incubated at rt for 120 min, then stopped by addition of 3 ⁇ _ per of quench solution (2.5% TFA with 320 nM d4-SAH).
  • the reaction mixture was centrifuged (Eppendorf centrifuge 5810, Rotor A-4-62) for 2 min at 2000 rpm and read on an API 4000 triple quadrupole mass spec with Turbulon Spray (Applied Biosystem) coupled with Prominence UFLC (Shimadzu).
  • the levels of SAH production were then normalized based on the values coming from the positive and negative controls to give percent enzyme activities.
  • the data were then fit to a dose response equation using the program Helios to get the IC 50 values of the test compound.
  • Representative compounds of the present disclosure were serially and separately diluted 3-fold in DMSO to obtain a total of eight or twelve concentrations.
  • the plate was gently agitated at 4°C for 30 min.
  • the cell lysate was neutralized with neutralization buffer (0.5 M sodium phosphate dibasic, pH 12.5, 1 mM DTT; 36 ⁇ _ per well).
  • neutralization buffer 0.5 M sodium phosphate dibasic, pH 12.5, 1 mM DTT; 36 ⁇ _ per well. The plate was agitated to ensure the lysates were well mixed prior to the ELISA protocol.
  • ELISA protocol Cell lysates were transferred to the wells of a 384-well plate and the final volume was adjusted to 50 ⁇ _ per well with PBS. The plate was sealed, centrifuged at 2,000 rpm for 2 min and incubated at 4°C for about 16 h. The plate was washed with TBST buffer (1 x TBS (1 Ox TBS: 24.2 g Tris (Sigma, T6066), 80 g NaCI (Sigma, S3014) to 1 L of water and adjust pH to 7.6 with HCI) with 0.1 % Tween-20). Blocking buffer (TBST, 5% BSA; 50 ⁇ _ per well) was added and the plate was incubated for 1 h at rt.
  • the blocking buffer was removed and primary antibody was added (30 ⁇ _ per well).
  • the following dilutions were performed with blocking buffer: for anti- H3K27me3 antibody (Cell Signaling Technology, #9733), dilution was 1 :1000; for anti- H3K27me2 antibody (Cell Signaling Technology, #9288), dilution was 1 :100; for anti-H3 antibody (Abeam, Cat#24834), dilution was 1 :1000.
  • the primary antibody was incubated in the plate at rt for 1 h.
  • the wells were washed with TBST and incubated with secondary antibody for 1 h at rt.
  • Representative compounds of the present disclosure were analyzed for their ability to selectively inhibit PRC2.
  • Western blot was performed using standard molecular biology techniques. Cell was lysed in SDS lysis buffer (Millipore, Cat#20-163) and protein concentration was measured by BCA protein assay (Pierce, Cat# PI-23221).
  • Antibodies for western blots were purchased from Cell Signaling Technology (Danvers, MA, USA).
  • Anti-H3K9me1 (#07- 395), anti-H3K27me1 (#07-448), and anti-H3K36me1 (#07-548) were purchased from Millipore (Billerica, MA, USA). Anti-H3K36me3 (ab9050-100) was purchased from Abeam (Cambridge, UK). Anti-H3K9me3 (#39161) was purchased from Active Motif (Carlsbad, CA, USA).
  • Compounds of the present disclosure specifically inhibit the methylation of the PRC2 substrate H3K27. This can be demonstrated by their ability to inhibit H3K27me2 and H3K27me3 in a number of human cancer cell lines, examples include rhabdoid cells (G401) and lymphoma cells (WSU-DLCL2, KARPAS422, SU-DHL4). Selectivity is profiled against a number of other methylation marks, for example: H3K4me2;
  • H3K9me2 H3K36me3; and H3K79me3.
  • B cell lymphoma cell KARPAS422 was cultured using standard cell culture conditions in RPMI-1640 (Invitrogen, cat #1 1875) supplemented with 15% FBS
  • a representative compound of the present disclosure was dissolved in 10% PEG300, 10% Solutol HS 15 and 80% pH 4.65 Acetate buffer to yield a final concentration of 0.2 mg/mL for intravenous (IV) and oral administration (PO).
  • rat PK studies a total of three male Sprague Dawley rats each were used for rat IV and PO PK study, respectively.
  • the formulation solution was administered via a single bolus IV at 1 mg/kg and a single oral gavage (PO) at 2 mg/kg, respectively.
  • Blood samples (approximately 150 ⁇ _) were collected via jugular cannula at appropriate time points.
  • PK parameters including area under concentration curve (AUC), mean residence time (MRT), plasma clearance (CI), steady state volume of distribution (Vdss), elimination half-life (t 1/2 ), maximum concentration (Cmax), time of maximum concentration (Tmax) and oral bioavailability (F %) were calculated using the following equations:
  • t time and C is plasma concentration at the time (t);
  • Dose iv is the dose for intravenous administration; and Dose ora i is the dose for oral administration.
  • Vdss CI * MRT
  • the cells Karpas 422 human B cell lymphoma were cultured in RPMI-1640 medium (Gibco; 1 1875-093) supplemented with 15% FBS (Gibco; 10099-141 ) and 1 % Pen Strep (Gibco; 15140-122) at 37°C in an atmosphere of 5% C02 in air. Cells were maintained in suspension cultures at concentrations between 0.5 - 2 x 10 6 cells/ml. Cells were split at 1 :3 every 2-4 days.
  • the cells were collected, suspended in PBS, mixed with Matrigel (BD Bioscience) at a volume ratio of 1 :1 at a concentration of 1 x10 8 cells/mL and then injected subcutaneously into the right flank of balb/c nude mice (Vital River) at a concentration of 5x10 6 cells per animal.
  • Matrigel BD Bioscience
  • the compound was formulated as a suspension in 0.5% methyl cellulose (MC) and 0.5% Tween 80 in 50 mM pH6.8 buffer (prepared in house according to the USP) and administered orally by gavage at specific doses.
  • MC methyl cellulose
  • Tween 80 in 50 mM pH6.8 buffer (prepared in house according to the USP) and administered orally by gavage at specific doses.
  • Treatment was initiated when the average tumor volume reached 100-300 mm 3 .
  • Tumor growth and body weights were monitored at regular intervals.
  • the two largest diameters, width (W) and length (L), of the xenograft tumors were measured manually with calipers and the tumor volume was estimated using the formula: 0.5 x L x W 2 .
  • results are presented as mean ⁇ SEM. Graphing and statistical analysis was performed using GraphPad Prism 5.00 (GraphPad Software). Tumor and body weight change data were analyzed statistically. If the variances in the data were normally distributed (Bartlett's test for equal variances), the data were analyzed using one-way ANOVA with post hoc Dunnet's test for comparison of treatment versus control group. The post hoc Tukey test was used for intragroup comparison. Otherwise, the Kruskal-Wallis ranked test post hoc Dunn's was used.
  • Tumor regression was calculated according to:
  • Atumor volumes represent the mean tumor volume on the evaluation day minus the mean tumor volume at the start of the experiment.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
PCT/IB2015/059843 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof WO2016103155A1 (en)

Priority Applications (31)

Application Number Priority Date Filing Date Title
MEP-2019-127A ME03385B (me) 2014-12-23 2015-12-21 Triazolopirimidin jedinjenja i njihova upotreba
JP2017533767A JP6736559B2 (ja) 2014-12-23 2015-12-21 トリアゾロピリミジン化合物およびその使用
SI201530725T SI3237418T1 (sl) 2014-12-23 2015-12-21 Spojine triazolopirimidina in njihove uporabe
RS20190502A RS58679B1 (sr) 2014-12-23 2015-12-21 Triazolopirimidin jednijenja i njihova upotreba
NZ731664A NZ731664A (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
CA2969090A CA2969090C (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
LTEP15820637.5T LT3237418T (lt) 2014-12-23 2015-12-21 Triazolpirimidino junginiai ir jų panaudojimas
EA201791420A EA032416B1 (ru) 2014-12-23 2015-12-21 Соединения триазолопиримидина и их применения
TN2017000204A TN2017000204A1 (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
DK15820637.5T DK3237418T3 (da) 2014-12-23 2015-12-21 Triazolopyrimidinforbindelser og anvendelser deraf
MX2017008529A MX2017008529A (es) 2014-12-23 2015-12-21 Compuestos de triazolopirimidina y usos de los mismos.
KR1020177016880A KR102534028B1 (ko) 2014-12-23 2015-12-21 트리아졸로피리미딘 화합물 및 그의 용도
ES15820637T ES2722048T3 (es) 2014-12-23 2015-12-21 Compuestos de triazolopirimidina y usos de los mismos
BR112017010354A BR112017010354B8 (pt) 2014-12-23 2015-12-21 Compostos de triazolopirimidina, seu uso, e composição farmacêutica
CR20170285A CR20170285A (es) 2014-12-23 2015-12-21 Compuestos de triazolopirimidina y usos de los mismos
AU2015370524A AU2015370524B2 (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
EP15820637.5A EP3237418B1 (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
UAA201704770A UA120945C2 (uk) 2014-12-23 2015-12-21 Сполуки триазолопіримідину та їх застосування
SG11201703880VA SG11201703880VA (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
US15/538,348 US10220036B2 (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
CN201580070691.5A CN107108637B (zh) 2014-12-23 2015-12-21 三唑并嘧啶化合物及其用途
MYPI2017701623A MY186837A (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
PL15820637T PL3237418T3 (pl) 2014-12-23 2015-12-21 Związki triazolopirymidyny i ich zastosowania
IL252135A IL252135B (en) 2014-12-23 2017-05-07 Triazolopyrimidine compounds and their uses
PH12017501016A PH12017501016B1 (en) 2014-12-23 2017-06-01 Triazolopyrimidine compounds and uses thereof
CONC2017/0005992A CO2017005992A2 (es) 2014-12-23 2017-06-16 Compuestos de triazolopirimidina y usos de los mismos
US16/246,380 US20190142837A1 (en) 2014-12-23 2019-01-11 Triazolopyrimidine compounds and uses thereof
HRP20190805TT HRP20190805T1 (hr) 2014-12-23 2019-04-29 Spojevi triazolopirimidina i njihova uporaba
CY20191100456T CY1121901T1 (el) 2014-12-23 2019-04-30 Ενωσεις τριαζολοπυριμιδινης και χρησεις αυτων
US16/863,631 US11207325B2 (en) 2014-12-23 2020-04-30 Triazolopyrimidine compounds and uses thereof
US17/505,271 US11931363B2 (en) 2014-12-23 2021-10-19 Triazolopyrimidine compounds and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CNPCT/CN2014/094644 2014-12-23
CN2014094644 2014-12-23
CNPCT/CN2015/095320 2015-11-23
CN2015095320 2015-11-23

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/538,348 A-371-Of-International US10220036B2 (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof
US16/246,380 Continuation US20190142837A1 (en) 2014-12-23 2019-01-11 Triazolopyrimidine compounds and uses thereof

Publications (1)

Publication Number Publication Date
WO2016103155A1 true WO2016103155A1 (en) 2016-06-30

Family

ID=55071101

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2015/059843 WO2016103155A1 (en) 2014-12-23 2015-12-21 Triazolopyrimidine compounds and uses thereof

Country Status (40)

Country Link
US (5) US10220036B2 (ko)
EP (1) EP3237418B1 (ko)
JP (1) JP6736559B2 (ko)
KR (1) KR102534028B1 (ko)
CN (1) CN107108637B (ko)
AU (1) AU2015370524B2 (ko)
BR (1) BR112017010354B8 (ko)
CA (1) CA2969090C (ko)
CL (1) CL2017001572A1 (ko)
CO (1) CO2017005992A2 (ko)
CR (1) CR20170285A (ko)
CY (1) CY1121901T1 (ko)
DK (1) DK3237418T3 (ko)
DO (1) DOP2017000149A (ko)
EA (1) EA032416B1 (ko)
EC (1) ECSP17047153A (ko)
ES (1) ES2722048T3 (ko)
GT (1) GT201700146A (ko)
HR (1) HRP20190805T1 (ko)
HU (1) HUE043060T2 (ko)
IL (1) IL252135B (ko)
JO (1) JO3489B1 (ko)
LT (1) LT3237418T (ko)
ME (1) ME03385B (ko)
MX (1) MX2017008529A (ko)
MY (1) MY186837A (ko)
NZ (1) NZ731664A (ko)
PE (1) PE20171307A1 (ko)
PH (1) PH12017501016B1 (ko)
PL (1) PL3237418T3 (ko)
PT (1) PT3237418T (ko)
RS (1) RS58679B1 (ko)
SG (1) SG11201703880VA (ko)
SI (1) SI3237418T1 (ko)
SV (1) SV2017005472A (ko)
TN (1) TN2017000204A1 (ko)
TW (1) TWI694076B (ko)
UA (1) UA120945C2 (ko)
UY (1) UY36462A (ko)
WO (1) WO2016103155A1 (ko)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017219948A1 (en) * 2016-06-20 2017-12-28 Novartis Ag Crystalline forms of triazolopyrimidine compound
WO2017221092A1 (en) * 2016-06-20 2017-12-28 Novartis Ag Triazolopyridine compounds and uses thereof
WO2019062435A1 (zh) * 2017-09-28 2019-04-04 中国科学院上海药物研究所 三氮唑并嘧啶、三氮唑并吡啶化合物及其组合物用于治疗prc2介导的疾病
US10266542B2 (en) 2017-03-15 2019-04-23 Mirati Therapeutics, Inc. EZH2 inhibitors
WO2019120276A1 (zh) * 2017-12-21 2019-06-27 上海青煜医药科技有限公司 嘧啶酮化合物及其应用
WO2019145176A1 (en) * 2018-01-23 2019-08-01 Basf Se Halogenation of pyridine derivatives
JP2019524872A (ja) * 2016-06-20 2019-09-05 ノバルティス アーゲー 癌の治療に有用なイミダゾピリミジン化合物
CN110734436A (zh) * 2018-07-19 2020-01-31 上海青煜医药科技有限公司 嘧啶或吡嗪并环化合物及其应用
WO2020156479A1 (zh) * 2019-02-02 2020-08-06 上海青煜医药科技有限公司 环丙烯并苯并呋喃取代的氮杂芳基化合物、其中间体、制备方法及应用
CN112004816A (zh) * 2018-01-31 2020-11-27 米拉蒂医疗股份有限公司 Prc2抑制剂
JP2020535215A (ja) * 2017-09-28 2020-12-03 中国科学院上海薬物研究所Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Prc2介在の疾患を治療するためのトリアゾロピリミジン、トリアゾロピリジン化合物及びその組成物
WO2020247475A1 (en) * 2019-06-05 2020-12-10 Mirati Therapeutics, Inc. Imidazo[1,2-c]pyrimidine derivatives as prc2 inhibitors for treating cancer
JP2021506776A (ja) * 2017-12-15 2021-02-22 レヴォリューション・メディスンズ,インコーポレイテッド アロステリックshp2阻害剤としての多環式化合物
WO2021057853A1 (en) * 2019-09-26 2021-04-01 Novartis Ag Aza-quinoline compounds and uses thereof
WO2021083380A1 (zh) * 2019-11-01 2021-05-06 上海科技大学 Eed抑制剂及其制备方法和用途
CN113004233A (zh) * 2019-12-18 2021-06-22 南京优氟医药科技有限公司 一种用于制备prc2抑制剂的化合物、其制备方法和用途
CN113636993A (zh) * 2020-05-11 2021-11-12 苏州亚盛药业有限公司 (5-氟-2,3-二氢苯并呋喃-4-基)甲胺或其盐的制备方法及其中间体
WO2021240373A1 (en) 2020-05-28 2021-12-02 Novartis Ag Dosing regimens for n-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-8-(2-methylpyridin-3-yl)-[1,2,4]triazolo[4,3-c]pyrimidin-5-amine, or a pharmaceutically acceptable salt thereof, for use in treating prc2-mediated diseases or disorders
CN114144230A (zh) * 2019-03-15 2022-03-04 弗尔康医疗公司 作为eed和prc2调节剂的大环唑并吡啶衍生物
WO2023016511A1 (zh) * 2021-08-11 2023-02-16 上海青煜医药科技有限公司 氮杂芳基化合物、其制备方法及应用
US11661401B2 (en) 2016-07-12 2023-05-30 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric SHP2 inhibitors
US11673896B2 (en) 2017-01-23 2023-06-13 Revolution Medicines, Inc. Pyridine compounds as allosteric SHP2 inhibitors
US11702411B2 (en) 2017-10-12 2023-07-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors
US11739093B2 (en) 2017-01-23 2023-08-29 Revolution Medicines, Inc. Substituted pyrazolopyrazines, imidazopyrazines and [1,2,4]triazolopyrazines as allosteric SHP2 inhibitors

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201703880VA (en) * 2014-12-23 2017-07-28 Novartis Ag Triazolopyrimidine compounds and uses thereof
JP7121660B2 (ja) 2016-06-01 2022-08-18 エピザイム,インコーポレイティド 癌を処置するためのezh2阻害剤の使用
CN106831601A (zh) * 2017-01-13 2017-06-13 河北博伦特药业有限公司 一种2‑氨基甲基嘧啶盐酸盐及其衍生物的合成方法
WO2018170290A1 (en) * 2017-03-15 2018-09-20 Fulcrum Therapeutics, Inc. Compositions and methods for increasing fmr1 expression
US11312705B2 (en) * 2017-03-16 2022-04-26 Jiangsu Hengrui Medicine Co., Ltd. Heteroaryl[4,3-C]pyrimidine-5-amine derivative, preparation method therefor, and medical uses thereof
WO2018223030A1 (en) 2017-06-02 2018-12-06 Epizyme, Inc. Use of ezh2 inhibitors for treating cancer
CN109867645B (zh) * 2017-12-05 2021-09-14 中国医学科学院药物研究所 一种2,2-二氟-2,3-二氢取代苯并呋喃类化合物的合成方法
CN110156787B (zh) * 2018-02-13 2021-11-02 中国科学院上海药物研究所 一种三氮唑并嘧啶衍生化合物、包含其的药物组合物及其用途
CN110563722A (zh) * 2018-06-06 2019-12-13 上海青煜医药科技有限公司 吡啶或哒嗪并环化合物及其应用
MX2021006026A (es) * 2018-11-30 2021-07-06 Tuojie Biotech Shanghai Co Ltd Pirimidina y derivado heterociclico de nitrogeno de cinco miembros, metodo de preparacion y usos medicos de los mismos.
WO2020204024A1 (ja) * 2019-04-03 2020-10-08 テラ・ストーン株式会社 チミン核酸塩基をベースとするトリアゾロピリミジン類及びその製造方法
EP3959214A1 (en) * 2019-04-22 2022-03-02 Mirati Therapeutics, Inc. Naphthyridine derivatives as prc2 inhibitors
CA3138307A1 (en) 2019-05-13 2020-11-19 Ecolab Usa Inc. 1,2,4-triazolo[1,5-a] pyrimidine derivative as copper corrosion inhibitor
AU2020288640A1 (en) * 2019-06-05 2021-12-23 Atnx Spv, Llc Methods of treating and/or preventing psoriasis
AU2020332462B2 (en) * 2019-08-22 2023-08-17 Shanghai Blueray Biopharma Co., Ltd. Azaheteroaryl compound and application thereof
CN111333655B (zh) * 2020-04-13 2021-07-13 武汉工程大学 一种三唑并嘧啶类化合物及其制备方法和应用
CN114907385A (zh) 2021-02-10 2022-08-16 上海青煜医药科技有限公司 氮杂芳基化合物、其制备方法及应用
CN117384153A (zh) * 2022-07-12 2024-01-12 上海赛岚生物科技有限公司 一类甲基转移酶抑制剂及其用途

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2779780A (en) 1955-03-01 1957-01-29 Du Pont 1, 4-diamino-2, 3-dicyano-1, 4-bis (substituted mercapto) butadienes and their preparation
US5621002A (en) 1993-09-09 1997-04-15 Behringwerke Aktiengesellschaft Prodrugs for enzyme mediated activation
WO2000035436A2 (en) 1998-12-16 2000-06-22 Warner-Lambert Company Treatment of arthritis with mek inhibitors
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2003044021A2 (en) * 2001-11-16 2003-05-30 Amgen Inc. Substituted indolizine-like compounds and methods of use
WO2003076424A1 (en) 2002-03-08 2003-09-18 Eisai Co. Ltd. Macrocyclic compounds useful as pharmaceuticals
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
WO2006122806A2 (en) 2005-05-20 2006-11-23 Novartis Ag 1,3-dihydro-imidazo [4,5-c] quinolin-2-ones as lipid kinase inhibitors
WO2007014011A2 (en) 2005-07-21 2007-02-01 Ardea Biosciences, Inc. N-(arylamino)-sulfonamide inhibitors of mek
WO2007084786A1 (en) 2006-01-20 2007-07-26 Novartis Ag Pyrimidine derivatives used as pi-3 kinase inhibitors
WO2009036082A2 (en) 2007-09-12 2009-03-19 Genentech, Inc. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents, and methods of use
WO2009055730A1 (en) 2007-10-25 2009-04-30 Genentech, Inc. Process for making thienopyrimidine compounds
WO2009155386A1 (en) 2008-06-20 2009-12-23 Abbott Laboratories A process for the preparation of the apoptosis promoter abt-263
WO2014144747A1 (en) * 2013-03-15 2014-09-18 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL1014106C2 (nl) 2000-01-18 2001-07-20 Univ Amsterdam Werkwijze voor het detecteren van een pathologische afwijking bij een cel, een antistof en een assay kit.
SE519662C2 (sv) 2001-07-04 2003-03-25 Asept Int Ab Ventilanordning vid utportioneringsanordningar
AU2002357667A1 (en) * 2001-10-24 2003-05-06 Iconix Pharmaceuticals, Inc. Modulators of phosphoinositide 3-kinase
GB0228900D0 (en) 2002-12-11 2003-01-15 Ml Lab Plc Cancer Immunotherapy
JP2004238296A (ja) 2003-02-04 2004-08-26 Kissei Pharmaceut Co Ltd 新規なトリアゾロピリミジン誘導体、それを含有する医薬組成物およびそれらの用途
WO2005005601A2 (en) 2003-06-09 2005-01-20 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US20060246505A1 (en) 2003-09-02 2006-11-02 Reinhard Walther Modulation of the synthesis of insulin
JP5160885B2 (ja) 2004-06-01 2013-03-13 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル 再構成ヒストンメチルトランスフェラーゼ複合体及びそのモジュレーターの識別方法
US7452892B2 (en) 2005-06-17 2008-11-18 Bristol-Myers Squibb Company Triazolopyrimidine cannabinoid receptor 1 antagonists
US7890267B2 (en) 2006-03-31 2011-02-15 Ordway Research Institute Prognostic and diagnostic method for cancer therapy
CA2673784A1 (en) 2006-12-27 2008-07-10 Usc Stevens-University Of Southern California Dna methylation markers based on epigenetic stem cell signatures in cancer
WO2009017670A2 (en) 2007-07-26 2009-02-05 University Of Massachusetts Ras-mediated epigenetic silencing effectors and uses thereof
WO2009026720A1 (en) 2007-08-29 2009-03-05 Methylgene Inc. Processes and intermediates for preparing fused heterocyclic kinase inhibitors
GB0822248D0 (en) 2008-12-05 2009-01-14 Medical Res Council Crystal meths
WO2010111712A2 (en) 2009-03-27 2010-09-30 Cold Spring Harbor Laboratory Identification of rnai targets and use of rnai for rational therapy of chemotherapy-resistant leukemia and other cancers
CN102020643A (zh) 2009-09-22 2011-04-20 上海恒瑞医药有限公司 二氢喋啶酮类衍生物、其制备方法及其在医药上的应用
TW201200518A (en) 2010-03-10 2012-01-01 Kalypsys Inc Heterocyclic inhibitors of histamine receptors for the treatment of disease
SI2566327T1 (sl) * 2010-05-07 2017-07-31 Glaxosmithkline Llc Indoli
KR102373074B1 (ko) 2010-09-10 2022-03-11 에피자임, 인코포레이티드 인간 ezh2의 억제제 및 이의 사용 방법
WO2012151277A1 (en) 2011-05-02 2012-11-08 Applied Informatic Solutions, Inc. Kits and methods for selecting a treatment for ovarian cancer
WO2013010181A2 (en) 2011-07-14 2013-01-17 University Of Massachusetts Methods of diagnosing cancer using epigenetic biomarkers
US9730925B2 (en) 2011-09-30 2017-08-15 Glaxomithkline Llc Methods of treating cancer
KR102587175B1 (ko) 2012-03-12 2023-10-06 에피자임, 인코포레이티드 인간 ezh2의 억제제 및 이의 사용 방법
WO2014078813A1 (en) 2012-11-19 2014-05-22 Irm Llc Compounds and compositions for the treatment of parasitic diseases
EP2934531A4 (en) 2012-12-19 2016-06-15 Glaxosmithkline Llc COMBINATION
US10047163B2 (en) 2013-02-08 2018-08-14 Abbvie Stemcentrx Llc Multispecific constructs
SG11201506077XA (en) 2013-02-11 2015-08-28 Constellation Pharmaceuticals Inc Modulators of methyl modifying enzymes, compositions and uses thereof
RU2015139054A (ru) 2013-03-14 2017-04-19 Дженентек, Инк. Способы лечения рака и профилактики лекарственной резистентности рака
WO2014151142A1 (en) 2013-03-15 2014-09-25 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US10167286B2 (en) 2014-02-11 2019-01-01 Mitokinin, Inc. Compositions and methods using the same for treatment of neurodegenerative and mitochondrial disease
HUE042290T2 (hu) 2014-11-14 2019-06-28 Basf Se Benzil-propargil-éterek, mint nitrifikáció inhibitorok
ES2668688T3 (es) 2014-12-16 2018-05-21 Omya International Ag Carbonato de calcio para protección vegetal
SG11201703880VA (en) * 2014-12-23 2017-07-28 Novartis Ag Triazolopyrimidine compounds and uses thereof
GB201501017D0 (en) 2014-12-23 2015-03-04 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against hepatocellular carcinoma (HCC) and other cancers
MA41272A (fr) 2014-12-23 2017-10-31 Adama Makhteshim Ltd 5-fluoro-4-imino-3-(alkyle/alkyle substitué)-1-(arylsulfonyl)-3,4-dihydropyrimidin-2(1h)-one en tant que traitement des semences

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2779780A (en) 1955-03-01 1957-01-29 Du Pont 1, 4-diamino-2, 3-dicyano-1, 4-bis (substituted mercapto) butadienes and their preparation
US5621002A (en) 1993-09-09 1997-04-15 Behringwerke Aktiengesellschaft Prodrugs for enzyme mediated activation
WO2000035436A2 (en) 1998-12-16 2000-06-22 Warner-Lambert Company Treatment of arthritis with mek inhibitors
WO2002006213A2 (en) 2000-07-19 2002-01-24 Warner-Lambert Company Oxygenated esters of 4-iodo phenylamino benzhydroxamic acids
WO2003044021A2 (en) * 2001-11-16 2003-05-30 Amgen Inc. Substituted indolizine-like compounds and methods of use
WO2003076424A1 (en) 2002-03-08 2003-09-18 Eisai Co. Ltd. Macrocyclic compounds useful as pharmaceuticals
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
WO2006122806A2 (en) 2005-05-20 2006-11-23 Novartis Ag 1,3-dihydro-imidazo [4,5-c] quinolin-2-ones as lipid kinase inhibitors
WO2007014011A2 (en) 2005-07-21 2007-02-01 Ardea Biosciences, Inc. N-(arylamino)-sulfonamide inhibitors of mek
WO2007084786A1 (en) 2006-01-20 2007-07-26 Novartis Ag Pyrimidine derivatives used as pi-3 kinase inhibitors
WO2009036082A2 (en) 2007-09-12 2009-03-19 Genentech, Inc. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents, and methods of use
WO2009055730A1 (en) 2007-10-25 2009-04-30 Genentech, Inc. Process for making thienopyrimidine compounds
WO2009155386A1 (en) 2008-06-20 2009-12-23 Abbott Laboratories A process for the preparation of the apoptosis promoter abt-263
WO2014144747A1 (en) * 2013-03-15 2014-09-18 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
"Beilsteins Handbuch der organischen Chemie", SPRINGER-VERLAG
"Remington: The Science and Practice of Pharmacy", 2012, PHARMACEUTICAL PRESS
ALLEN, L.V., JR. ET AL.: "Remington: The Science and Practice of Pharmacy", vol. 2, 2012, PHARMACEUTICAL PRESS
BAI, J ET AL., CELL PROLIF., vol. 47, no. 3, 2014, pages 211 - 218
BAI, J. ET AL., CELL PROLIF., vol. 47, no. 3, 2014, pages 211 - 218
BEGUELIN, W. ET AL., CANCER CELL, vol. 23, no. 5, 2013, pages 677 - 692
BHAN, A. ET AL., J MOL BIOL., 2014
CHANG, C. ET AL., CANCER CELL, vol. 19, no. 1, 2011, pages 86 - 100
CHANG, C. J. ET AL., CANCER CELL, vol. 19, no. 1, 2011, pages 86 - 100
CHEN B. ET AL., CELL CYCLE, vol. 12, no. 1, 2013, pages 112 - 121
CHEN, S ET AL., NAT CELL BIOL., vol. 12, no. 11, 2010, pages 1108 - 1114
FORMENKO, I. ET AL., OMPUTER METHODS AND PROGRAMS IN BIOMEDICINE, vol. 82, 2006, pages 31 - 37
GONZALEZ, M ET AL., CANCER RES., vol. 71, no. 6, 2011, pages 2360 - 2370
GREENE, T.W ET AL.: "Protecting Groups in Organic Synthesis", 2007, WILEY
HEBBARD, L., ONCOGENE, vol. 30, no. 3, 2011, pages 301 - 312
KAHM, M ET AL., JOURNAL OF STATISTICAL SOFTWARE, vol. 33, no. 7, 2010
KELLY, C ET AL., BIOMETRICS, vol. 46, no. 4, 1990, pages 1071 - 1085
KNUTSON, S. ET AL., PLOS ONE, 2014
LAROCK, R.C.: "Comprehensive Organic Transformations", 1999, WILEY-VCH
LOUIS F. FIESER; MARY FIESER: "Reagents for Organic Synthesis", vol. 1-19, 1967, WILEY
MUSCH, T. ET AL., PLOS ONE, 2010, pages E10726
NORMOLLE, D. P., STATISTICS IN MEDICINE, vol. 12, 1993, pages 2025 - 2042
PATHIRAJA, T. ET AL., SCI. TRANSL. MED., vol. 6, no. 229, 2014, pages 229 RA41
ROJANASAKUL, Y., CELL CYCLE, vol. 12, no. 2, 2013, pages 202 - 203
SEBAUGH, J. L., PHARMACEUTICAL STATISTICS, vol. 10, 2011, pages 128 - 134
WU, Z. ET AL., CELL DEATH DIFFER., vol. 18, no. 11, 2011, pages 1771 - 1779
YAMAGUCHI, J ET AL., CANCER SCI., vol. 101, no. 2, 2010, pages 355 - 362
ZENG, X. ET AL., CELL CYCLE, vol. 10, no. 4, 2011, pages 579 - 583

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2017282871B2 (en) * 2016-06-20 2020-03-05 Novartis Ag Crystalline forms of triazolopyrimidine compound
CN109563100A (zh) * 2016-06-20 2019-04-02 诺华股份有限公司 三唑并嘧啶化合物的晶体形式
JP2019524872A (ja) * 2016-06-20 2019-09-05 ノバルティス アーゲー 癌の治療に有用なイミダゾピリミジン化合物
EP3472168A4 (en) * 2016-06-20 2019-12-11 Novartis AG CRYSTALLINE FORMS OF A TRIAZOLOPYRIMIDINE COMPOUND
JP2019522049A (ja) * 2016-06-20 2019-08-08 ノバルティス アーゲー トリアゾロピリジン化合物及びその使用
CN109906224A (zh) * 2016-06-20 2019-06-18 诺华股份有限公司 三唑吡啶化合物及其应用
CN109563100B (zh) * 2016-06-20 2021-08-27 诺华股份有限公司 三唑并嘧啶化合物的晶体形式
CN109906224B (zh) * 2016-06-20 2022-02-25 诺华股份有限公司 三唑吡啶化合物及其应用
US11091489B2 (en) 2016-06-20 2021-08-17 Novartis Ag Crystalline forms of a triazolopyrimidine compound
WO2017221092A1 (en) * 2016-06-20 2017-12-28 Novartis Ag Triazolopyridine compounds and uses thereof
US10689378B2 (en) 2016-06-20 2020-06-23 Novartis Ag Triazolopyridine compounds and uses thereof
US11548897B2 (en) 2016-06-20 2023-01-10 Novartis Ag Crystalline forms of a triazolopyrimidine compound
WO2017219948A1 (en) * 2016-06-20 2017-12-28 Novartis Ag Crystalline forms of triazolopyrimidine compound
US10676479B2 (en) 2016-06-20 2020-06-09 Novartis Ag Imidazolepyridine compounds and uses thereof
US11661401B2 (en) 2016-07-12 2023-05-30 Revolution Medicines, Inc. 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric SHP2 inhibitors
US11739093B2 (en) 2017-01-23 2023-08-29 Revolution Medicines, Inc. Substituted pyrazolopyrazines, imidazopyrazines and [1,2,4]triazolopyrazines as allosteric SHP2 inhibitors
US11673896B2 (en) 2017-01-23 2023-06-13 Revolution Medicines, Inc. Pyridine compounds as allosteric SHP2 inhibitors
US10266542B2 (en) 2017-03-15 2019-04-23 Mirati Therapeutics, Inc. EZH2 inhibitors
US11013745B2 (en) 2017-09-28 2021-05-25 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Triazolopyrimidine, triazolopyridine compounds, and the composition thereof for treating PRC2-mediated diseases
WO2019062435A1 (zh) * 2017-09-28 2019-04-04 中国科学院上海药物研究所 三氮唑并嘧啶、三氮唑并吡啶化合物及其组合物用于治疗prc2介导的疾病
JP2020535215A (ja) * 2017-09-28 2020-12-03 中国科学院上海薬物研究所Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Prc2介在の疾患を治療するためのトリアゾロピリミジン、トリアゾロピリジン化合物及びその組成物
US11702411B2 (en) 2017-10-12 2023-07-18 Revolution Medicines, Inc. Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors
JP2021506776A (ja) * 2017-12-15 2021-02-22 レヴォリューション・メディスンズ,インコーポレイテッド アロステリックshp2阻害剤としての多環式化合物
JP7361693B2 (ja) 2017-12-15 2023-10-16 レヴォリューション・メディスンズ,インコーポレイテッド アロステリックshp2阻害剤としての多環式化合物
US11673901B2 (en) 2017-12-15 2023-06-13 Revolution Medicines, Inc. Polycyclic compounds as allosteric SHP2 inhibitors
WO2019120276A1 (zh) * 2017-12-21 2019-06-27 上海青煜医药科技有限公司 嘧啶酮化合物及其应用
US11591312B2 (en) 2018-01-23 2023-02-28 Basf Se Halogenation of pyridine derivatives
WO2019145176A1 (en) * 2018-01-23 2019-08-01 Basf Se Halogenation of pyridine derivatives
US11091495B2 (en) 2018-01-31 2021-08-17 Mirati Therapeutics, Inc. Substituted imidazo[1,2-c]pyrimidines as PRC2 inhibitors
US11220509B2 (en) 2018-01-31 2022-01-11 Mirati Therapeutics, Inc. Substituted imidazo[1,2-c]pyrimidines as PRC2 inhibitors
CN112004816B (zh) * 2018-01-31 2024-01-05 米拉蒂医疗股份有限公司 Prc2抑制剂
US11485738B2 (en) 2018-01-31 2022-11-01 Mirati Therapeutics, Inc. Substituted imidazo[1,2-c]pyrimidines as PRC2 inhibitors
CN112004816A (zh) * 2018-01-31 2020-11-27 米拉蒂医疗股份有限公司 Prc2抑制剂
CN110734436A (zh) * 2018-07-19 2020-01-31 上海青煜医药科技有限公司 嘧啶或吡嗪并环化合物及其应用
CN111518100A (zh) * 2019-02-02 2020-08-11 上海青煜医药科技有限公司 环丙烯并苯并呋喃取代的氮杂芳基化合物及其应用
WO2020156479A1 (zh) * 2019-02-02 2020-08-06 上海青煜医药科技有限公司 环丙烯并苯并呋喃取代的氮杂芳基化合物、其中间体、制备方法及应用
CN114144230B (zh) * 2019-03-15 2024-04-23 弗尔康医疗公司 作为eed和prc2调节剂的大环唑并吡啶衍生物
CN114144230A (zh) * 2019-03-15 2022-03-04 弗尔康医疗公司 作为eed和prc2调节剂的大环唑并吡啶衍生物
WO2020247475A1 (en) * 2019-06-05 2020-12-10 Mirati Therapeutics, Inc. Imidazo[1,2-c]pyrimidine derivatives as prc2 inhibitors for treating cancer
WO2021057853A1 (en) * 2019-09-26 2021-04-01 Novartis Ag Aza-quinoline compounds and uses thereof
WO2021083380A1 (zh) * 2019-11-01 2021-05-06 上海科技大学 Eed抑制剂及其制备方法和用途
EP4056570A4 (en) * 2019-11-01 2023-12-20 ShanghaiTech University EED INHIBITOR AND PRODUCTION METHOD AND USE THEREOF
CN114929707A (zh) * 2019-11-01 2022-08-19 上海科技大学 Eed抑制剂及其制备方法和用途
CN114929707B (zh) * 2019-11-01 2024-04-09 上海科技大学 Eed抑制剂及其制备方法和用途
CN113004233A (zh) * 2019-12-18 2021-06-22 南京优氟医药科技有限公司 一种用于制备prc2抑制剂的化合物、其制备方法和用途
CN113004233B (zh) * 2019-12-18 2022-12-20 南京优氟医药科技有限公司 一种用于制备prc2抑制剂的化合物、其制备方法和用途
CN113636993A (zh) * 2020-05-11 2021-11-12 苏州亚盛药业有限公司 (5-氟-2,3-二氢苯并呋喃-4-基)甲胺或其盐的制备方法及其中间体
WO2021228034A1 (en) * 2020-05-11 2021-11-18 Ascentage Pharma (Suzhou) Co., Ltd. Preparation method of (5-fluoro-2, 3-dihydrobenzofuran-4-yl) methanamine or its salt, and intermediates thereof
WO2021240373A1 (en) 2020-05-28 2021-12-02 Novartis Ag Dosing regimens for n-((5-fluoro-2,3-dihydrobenzofuran-4-yl)methyl)-8-(2-methylpyridin-3-yl)-[1,2,4]triazolo[4,3-c]pyrimidin-5-amine, or a pharmaceutically acceptable salt thereof, for use in treating prc2-mediated diseases or disorders
WO2023016511A1 (zh) * 2021-08-11 2023-02-16 上海青煜医药科技有限公司 氮杂芳基化合物、其制备方法及应用

Also Published As

Publication number Publication date
PE20171307A1 (es) 2017-09-05
US10220036B2 (en) 2019-03-05
TW201629065A (zh) 2016-08-16
CR20170285A (es) 2017-08-21
MY186837A (en) 2021-08-25
CA2969090A1 (en) 2016-06-30
PL3237418T3 (pl) 2019-07-31
EP3237418B1 (en) 2019-01-30
UY36462A (es) 2016-07-29
CL2017001572A1 (es) 2018-01-12
CY1121901T1 (el) 2020-10-14
US11931363B2 (en) 2024-03-19
CN107108637B (zh) 2019-10-29
TN2017000204A1 (en) 2018-10-19
BR112017010354B1 (pt) 2022-09-20
AU2015370524A1 (en) 2017-06-01
JP2018500342A (ja) 2018-01-11
EA201791420A1 (ru) 2017-10-31
ES2722048T3 (es) 2019-08-07
CA2969090C (en) 2023-05-02
US20170348312A1 (en) 2017-12-07
HUE043060T2 (hu) 2019-07-29
RS58679B1 (sr) 2019-06-28
PT3237418T (pt) 2019-05-23
US20230033320A1 (en) 2023-02-02
KR102534028B1 (ko) 2023-05-19
HRP20190805T1 (hr) 2019-06-28
DOP2017000149A (es) 2017-07-15
SI3237418T1 (sl) 2019-06-28
US20160176882A1 (en) 2016-06-23
SV2017005472A (es) 2018-06-12
TWI694076B (zh) 2020-05-21
US20190142837A1 (en) 2019-05-16
ME03385B (me) 2020-01-20
ECSP17047153A (es) 2019-02-28
IL252135B (en) 2020-06-30
BR112017010354A2 (pt) 2017-12-26
EP3237418A1 (en) 2017-11-01
GT201700146A (es) 2019-06-12
IL252135A0 (en) 2017-07-31
AU2015370524B2 (en) 2018-11-01
JO3489B1 (ar) 2020-07-05
JP6736559B2 (ja) 2020-08-05
DK3237418T3 (da) 2019-05-13
PH12017501016A1 (en) 2017-12-11
US20200323859A1 (en) 2020-10-15
BR112017010354B8 (pt) 2022-10-18
SG11201703880VA (en) 2017-07-28
LT3237418T (lt) 2019-05-10
PH12017501016B1 (en) 2017-12-11
US9580437B2 (en) 2017-02-28
CO2017005992A2 (es) 2017-10-20
UA120945C2 (uk) 2020-03-10
EA032416B1 (ru) 2019-05-31
US11207325B2 (en) 2021-12-28
KR20170095882A (ko) 2017-08-23
MX2017008529A (es) 2017-10-25
CN107108637A (zh) 2017-08-29
NZ731664A (en) 2024-02-23

Similar Documents

Publication Publication Date Title
US11931363B2 (en) Triazolopyrimidine compounds and uses thereof
US10676479B2 (en) Imidazolepyridine compounds and uses thereof
EP3472161B1 (en) Triazolopyridine compounds and uses thereof
JP6695353B2 (ja) Fgfr4阻害剤としてのホルミル化n−複素環式誘導体
WO2017149463A1 (en) Cyano-substituted indole compounds and uses thereof as lsd1 inhibitors
JP2022539208A (ja) チロシンキナーゼ非受容体1(tnk1)阻害剤およびその使用
EP4041725B1 (en) Quinoline compounds and compositions for inhibiting ezh2 as a treatment for cancer diseases

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15820637

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 252135

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2969090

Country of ref document: CA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112017010354

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2015370524

Country of ref document: AU

Date of ref document: 20151221

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12017501016

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 11201703880V

Country of ref document: SG

WWE Wipo information: entry into national phase

Ref document number: NC2017/0005992

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 20177016880

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15538348

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2017533767

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2017285

Country of ref document: CR

Ref document number: 001154-2017

Country of ref document: PE

Ref document number: MX/A/2017/008529

Country of ref document: MX

Ref document number: CR2017-000285

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: A201704770

Country of ref document: UA

REEP Request for entry into the european phase

Ref document number: 2015820637

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201791420

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 112017010354

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20170517